1
|
Radwan M, Guo T, Carvajal EG, Bekkema BAR, Cairo CW. Bioisosteres at C9 of 2-Deoxy-2,3-didehydro- N-acetyl Neuraminic Acid Identify Selective Inhibitors of NEU3. J Med Chem 2024; 67:13594-13603. [PMID: 39101748 DOI: 10.1021/acs.jmedchem.3c02186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
Human neuraminidases play critical roles in many physiological and pathological processes. Humans have four isoenzymes of NEU, making selective inhibitors important tools to investigate the function of individual isoenzymes. A typical scaffold for NEU inhibitors is 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (DANA) where C9 modifications can be critical for potency and selectivity against human NEU. To design improved DANA analogues, we generated a library of compounds with either a short alkyl chain or a biphenyl substituent linked to the C9 position through one of six amide bioisosteres. Bioisostere linkers included triazole, urea, thiourea, carbamate, thiocarbamate, and sulfonamide groups. Within this library, we identified a C9 biphenyl carbamate derivative (963) that showed high selectivity and potency for NEU3 (Ki = 0.12 ± 0.01 μM). In contrast, NEU1 and NEU4 isoenzymes preferred amide and triazole linkers, respectively. Finally, analogues with urea, sulfonamide, and amide linkers showed enhanced inhibitory activity for a bacterial NEU, NanI from Clostridium perfringens.
Collapse
Affiliation(s)
- Mostafa Radwan
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Tianlin Guo
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Elisa G Carvajal
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Benjamin A R Bekkema
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Christopher W Cairo
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| |
Collapse
|
2
|
Keil J, Rafn GR, Turan IM, Aljohani MA, Sahebjam-Atabaki R, Sun XL. Sialidase Inhibitors with Different Mechanisms. J Med Chem 2022; 65:13574-13593. [PMID: 36252951 PMCID: PMC9620260 DOI: 10.1021/acs.jmedchem.2c01258] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Indexed: 11/28/2022]
Abstract
Sialidases, or neuraminidases, are enzymes that catalyze the hydrolysis of sialic acid (Sia)-containing molecules, mostly removal of the terminal Sia (desialylation). By desialylation, sialidase can modulate the functionality of the target compound and is thus often involved in biological pathways. Inhibition of sialidases with inhibitors is an important approach for understanding sialidase function and the underlying mechanisms and could serve as a therapeutic approach as well. Transition-state analogues, such as anti-influenza drugs oseltamivir and zanamivir, are major sialidase inhibitors. In addition, difluoro-sialic acids were developed as mechanism-based sialidase inhibitors. Further, fluorinated quinone methide-based suicide substrates were reported. Sialidase product analogue inhibitors were also explored. Finally, natural products have shown competitive inhibiton against viral, bacterial, and human sialidases. This Perspective describes sialidase inhibitors with different mechanisms and their activities and future potential, which include transition-state analogue inhibitors, mechanism-based inhibitors, suicide substrate inhibitors, product analogue inhibitors, and natural product inhibitors.
Collapse
Affiliation(s)
- Joseph
M. Keil
- Department of Chemistry, Chemical and
Biomedical Engineering and Center for Gene Regulation in Health and
Disease (GRHD), Cleveland State University, Cleveland, Ohio 44115, United States
| | - Garrett R. Rafn
- Department of Chemistry, Chemical and
Biomedical Engineering and Center for Gene Regulation in Health and
Disease (GRHD), Cleveland State University, Cleveland, Ohio 44115, United States
| | - Isaac M. Turan
- Department of Chemistry, Chemical and
Biomedical Engineering and Center for Gene Regulation in Health and
Disease (GRHD), Cleveland State University, Cleveland, Ohio 44115, United States
| | - Majdi A. Aljohani
- Department of Chemistry, Chemical and
Biomedical Engineering and Center for Gene Regulation in Health and
Disease (GRHD), Cleveland State University, Cleveland, Ohio 44115, United States
| | - Reza Sahebjam-Atabaki
- Department of Chemistry, Chemical and
Biomedical Engineering and Center for Gene Regulation in Health and
Disease (GRHD), Cleveland State University, Cleveland, Ohio 44115, United States
| | - Xue-Long Sun
- Department of Chemistry, Chemical and
Biomedical Engineering and Center for Gene Regulation in Health and
Disease (GRHD), Cleveland State University, Cleveland, Ohio 44115, United States
| |
Collapse
|
3
|
Bourguet E, Figurska S, Fra Czek MM. Human Neuraminidases: Structures and Stereoselective Inhibitors. J Med Chem 2022; 65:3002-3025. [PMID: 35170942 DOI: 10.1021/acs.jmedchem.1c01612] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This Perspective describes the classification, structures, substrates, mechanisms of action, and implications of human neuraminidases (hNEUs) in various pathologies. Some inhibitors have been developed for each isoform, leading to more precise interactions with hNEUs. Although crystal structure data are available for NEU2, most of the findings are based on NEU1 inhibition, and limited information is available for other hNEUs. Therefore, the synthesis of new compounds would facilitate the enrichment of the arsenal of inhibitors to better understand the roles of hNEUs and their mechanisms of action. Nevertheless, due to the already known inhibitors of human neuraminidase enzymes, a structure-activity relationship is presented along with different approaches to inhibit these enzymes for the development of potent and selective inhibitors. Among the different emerging strategies, one is the inhibition of the dimerization of NEU1 or NEU3, and the second is the inhibition of certain receptors located close to hNEU.
Collapse
Affiliation(s)
- Erika Bourguet
- Université de Reims Champagne-Ardenne, Institut de Chimie Moléculaire de Reims (ICMR), CNRS UMR 7312, 51097 Reims, France
| | - Sylwia Figurska
- Université de Reims Champagne-Ardenne, Institut de Chimie Moléculaire de Reims (ICMR), CNRS UMR 7312, 51097 Reims, France.,Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Manuela Maria Fra Czek
- Université de Reims Champagne-Ardenne, Institut de Chimie Moléculaire de Reims (ICMR), CNRS UMR 7312, 51097 Reims, France.,Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| |
Collapse
|
4
|
Tembely D, Henry A, Vanalderwiert L, Toussaint K, Bennasroune A, Blaise S, Sartelet H, Jaisson S, Galés C, Martiny L, Duca L, Romier-Crouzet B, Maurice P. The Elastin Receptor Complex: An Emerging Therapeutic Target Against Age-Related Vascular Diseases. Front Endocrinol (Lausanne) 2022; 13:815356. [PMID: 35222273 PMCID: PMC8873114 DOI: 10.3389/fendo.2022.815356] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/20/2022] [Indexed: 12/26/2022] Open
Abstract
The incidence of cardiovascular diseases is increasing worldwide with the growing aging of the population. Biological aging has major influence on the vascular tree and is associated with critical changes in the morphology and function of the arterial wall together with an extensive remodeling of the vascular extracellular matrix. Elastic fibers fragmentation and release of elastin degradation products, also known as elastin-derived peptides (EDPs), are typical hallmarks of aged conduit arteries. Along with the direct consequences of elastin fragmentation on the mechanical properties of arteries, the release of EDPs has been shown to modulate the development and/or progression of diverse vascular and metabolic diseases including atherosclerosis, thrombosis, type 2 diabetes and nonalcoholic steatohepatitis. Most of the biological effects mediated by these bioactive peptides are due to a peculiar membrane receptor called elastin receptor complex (ERC). This heterotrimeric receptor contains a peripheral protein called elastin-binding protein, the protective protein/cathepsin A, and a transmembrane sialidase, the neuraminidase-1 (NEU1). In this review, after an introductive part on the consequences of aging on the vasculature and the release of EDPs, we describe the composition of the ERC, the signaling pathways triggered by this receptor, and the current pharmacological strategies targeting ERC activation. Finally, we present and discuss new regulatory functions that have emerged over the last few years for the ERC through desialylation of membrane glycoproteins by NEU1, and its potential implication in receptor transactivation.
Collapse
Affiliation(s)
- Dignê Tembely
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Aubéri Henry
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Laetitia Vanalderwiert
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Kevin Toussaint
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Amar Bennasroune
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Sébastien Blaise
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Hervé Sartelet
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Stéphane Jaisson
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Céline Galés
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse, Toulouse, France
| | - Laurent Martiny
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Béatrice Romier-Crouzet
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Pascal Maurice
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
- *Correspondence: Pascal Maurice, ; orcid.org0000-0003-2167-4808
| |
Collapse
|
5
|
Albrecht C, Akissi ZLE, Yao-Kouassi PA, Alabdul Magid A, Maurice P, Duca L, Voutquenne-Nazabadioko L, Bennasroune A. Identification and Evaluation of New Potential Inhibitors of Human Neuraminidase 1 Extracted from Olyra latifolia L.: A Preliminary Study. Biomedicines 2021; 9:biomedicines9040411. [PMID: 33920466 PMCID: PMC8070403 DOI: 10.3390/biomedicines9040411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 12/02/2022] Open
Abstract
Sialidases, also called neuraminidases, are involved in several human pathologies such as neurodegenerative disorders, cancers, as well as infectious and cardiovascular diseases. Several studies have shown that neuraminidases, such as neuraminidase 1 (NEU-1), may be promising pharmacological targets. Therefore, the discovery of new selective inhibitors of NEU-1 are necessary to better understand the biological functions of this sialidase. In the present study, we describe the isolation and characterization of nine known compounds from Olyra latifolia L. leaves. This plant, known to have several therapeutic properties, belongs to the family of Poaceae and is found in the neotropics and in tropical Africa and Madagascar. Among the purified compounds, feddeiketone B, 2,3-dihydroxy-1-(4-hydroxy-3,5-diméthoxyphényl)-l-propanone, and syringylglycerol were shown to present structural analogy with DANA, and their effects on membrane NEU-1 sialidase activity were evaluated. Our results show that they possess inhibitory effects against NEU-1-mediated sialidase activity at the plasma membrane. In conclusion, we identified new natural bioactive molecules extracted from Olyra latifolia as inhibitors of human NEU-1 of strong interest to elucidate the biological functions of this sialidase and to target this protein involved in several pathophysiological contexts.
Collapse
Affiliation(s)
- Camille Albrecht
- UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, CNRS, Université de Reims Champagne-Ardenne, 51097 Reims, France; (C.A.); (P.M.); (L.D.)
| | - Zachée Louis Evariste Akissi
- UMR 7312, Institut de Chimie Moléculaire de Reims (ICMR), UFR Sciences Exactes et Naturelles, CNRS, Université de Reims Champagne-Ardenne, 51097 Reims, France; (Z.L.E.A.); (A.A.M.)
- Laboratoire de Constitution et Réaction de la Matière, UFR Sciences des Structures de la Matière et de Technologie, Université Félix Houphouët-Boigny, 22 BP 582 Abidjan, Cote D’Ivoire;
| | - Philomène Akoua Yao-Kouassi
- Laboratoire de Constitution et Réaction de la Matière, UFR Sciences des Structures de la Matière et de Technologie, Université Félix Houphouët-Boigny, 22 BP 582 Abidjan, Cote D’Ivoire;
| | - Abdulmagid Alabdul Magid
- UMR 7312, Institut de Chimie Moléculaire de Reims (ICMR), UFR Sciences Exactes et Naturelles, CNRS, Université de Reims Champagne-Ardenne, 51097 Reims, France; (Z.L.E.A.); (A.A.M.)
| | - Pascal Maurice
- UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, CNRS, Université de Reims Champagne-Ardenne, 51097 Reims, France; (C.A.); (P.M.); (L.D.)
| | - Laurent Duca
- UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, CNRS, Université de Reims Champagne-Ardenne, 51097 Reims, France; (C.A.); (P.M.); (L.D.)
| | - Laurence Voutquenne-Nazabadioko
- UMR 7312, Institut de Chimie Moléculaire de Reims (ICMR), UFR Sciences Exactes et Naturelles, CNRS, Université de Reims Champagne-Ardenne, 51097 Reims, France; (Z.L.E.A.); (A.A.M.)
- Correspondence: (L.V.-N.); (A.B.)
| | - Amar Bennasroune
- UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, CNRS, Université de Reims Champagne-Ardenne, 51097 Reims, France; (C.A.); (P.M.); (L.D.)
- Correspondence: (L.V.-N.); (A.B.)
| |
Collapse
|
6
|
Albrecht C, Kuznetsov AS, Appert-Collin A, Dhaideh Z, Callewaert M, Bershatsky YV, Urban AS, Bocharov EV, Bagnard D, Baud S, Blaise S, Romier-Crouzet B, Efremov RG, Dauchez M, Duca L, Gueroult M, Maurice P, Bennasroune A. Transmembrane Peptides as a New Strategy to Inhibit Neuraminidase-1 Activation. Front Cell Dev Biol 2020; 8:611121. [PMID: 33392200 PMCID: PMC7772355 DOI: 10.3389/fcell.2020.611121] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/27/2020] [Indexed: 12/31/2022] Open
Abstract
Sialidases, or neuraminidases, are involved in several human disorders such as neurodegenerative, infectious and cardiovascular diseases, and cancers. Accumulative data have shown that inhibition of neuraminidases, such as NEU1 sialidase, may be a promising pharmacological target, and selective inhibitors of NEU1 are therefore needed to better understand the biological functions of this sialidase. In the present study, we designed interfering peptides (IntPep) that target a transmembrane dimerization interface previously identified in human NEU1 that controls its membrane dimerization and sialidase activity. Two complementary strategies were used to deliver the IntPep into cells, either flanked to a TAT sequence or non-tagged for solubilization in detergent micelles. Combined with molecular dynamics simulations and heteronuclear nuclear magnetic resonance (NMR) studies in membrane-mimicking environments, our results show that these IntPep are able to interact with the dimerization interface of human NEU1, to disrupt membrane NEU1 dimerization and to strongly decrease its sialidase activity at the plasma membrane. In conclusion, we report here new selective inhibitors of human NEU1 of strong interest to elucidate the biological functions of this sialidase.
Collapse
Affiliation(s)
- Camille Albrecht
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Andrey S Kuznetsov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Higher School of Economics, Moscow, Russia.,Moscow Institute of Physics and Technology, National Research University, Dolgoprudny, Russia
| | - Aline Appert-Collin
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Zineb Dhaideh
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Maïté Callewaert
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7312, Institut de Chimie Moléculaire de Reims, Reims, France
| | - Yaroslav V Bershatsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Moscow Institute of Physics and Technology, National Research University, Dolgoprudny, Russia
| | - Anatoly S Urban
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Moscow Institute of Physics and Technology, National Research University, Dolgoprudny, Russia
| | - Eduard V Bocharov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Moscow Institute of Physics and Technology, National Research University, Dolgoprudny, Russia
| | - Dominique Bagnard
- Université de Strasbourg, Strasbourg, France.,INSERM U1119 Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Stéphanie Baud
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Sébastien Blaise
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Béatrice Romier-Crouzet
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Roman G Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Higher School of Economics, Moscow, Russia.,Moscow Institute of Physics and Technology, National Research University, Dolgoprudny, Russia
| | - Manuel Dauchez
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France.,Plateau de Modélisation Moléculaire Multi-échelle, Reims, France
| | - Laurent Duca
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Marc Gueroult
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Pascal Maurice
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| | - Amar Bennasroune
- Université de Reims Champagne-Ardenne, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims, France
| |
Collapse
|
7
|
Karhadkar TR, Meek TD, Gomer RH. Inhibiting Sialidase-Induced TGF- β1 Activation Attenuates Pulmonary Fibrosis in Mice. J Pharmacol Exp Ther 2020; 376:106-117. [PMID: 33144389 DOI: 10.1124/jpet.120.000258] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
The active form of transforming growth factor-β1 (TGF-β1) plays a key role in potentiating fibrosis. TGF-β1 is sequestered in an inactive state by a latency-associated glycopeptide (LAP). Sialidases (also called neuraminidases (NEU)) cleave terminal sialic acids from glycoconjugates. The sialidase NEU3 is upregulated in fibrosis, and mice lacking Neu3 show attenuated bleomycin-induced increases in active TGF-β1 in the lungs and attenuated pulmonary fibrosis. Here we observe that recombinant human NEU3 upregulates active human TGF-β1 by releasing active TGF-β1 from its latent inactive form by desialylating LAP. Based on the proposed mechanism of action of NEU3, we hypothesized that compounds with a ring structure resembling picolinic acid might be transition state analogs and thus possible NEU3 inhibitors. Some compounds in this class showed nanomolar IC50 for recombinant human NEU3 releasing active human TGF-β1 from the latent inactive form. The compounds given as daily 0.1-1-mg/kg injections starting at day 10 strongly attenuated lung inflammation, lung TGF-β1 upregulation, and pulmonary fibrosis at day 21 in a mouse bleomycin model of pulmonary fibrosis. These results suggest that NEU3 participates in fibrosis by desialylating LAP and releasing TGF-β1 and that the new class of NEU3 inhibitors are potential therapeutics for fibrosis. SIGNIFICANCE STATEMENT: The extracellular sialidase NEU3 appears to be a key driver of pulmonary fibrosis. The significance of this report is that 1) we show the mechanism (NEU3 desialylates the latency-associated glycopeptide protein that keeps the profibrotic cytokine transforming growth factor-β1 (TGF-β1) in an inactive state, causing active TGF-β1 release), 2) we then use the predicted NEU3 mechanism to identify nM IC50 NEU3 inhibitors, and 3) these new NEU3 inhibitors are potent therapeutics in a mouse model of pulmonary fibrosis.
Collapse
Affiliation(s)
- Tejas R Karhadkar
- Departments of Biology (T.R.K., R.H.G.) and Biochemistry and Biophysics (T.D.M.), Texas A&M University, College Station, Texas
| | - Thomas D Meek
- Departments of Biology (T.R.K., R.H.G.) and Biochemistry and Biophysics (T.D.M.), Texas A&M University, College Station, Texas
| | - Richard H Gomer
- Departments of Biology (T.R.K., R.H.G.) and Biochemistry and Biophysics (T.D.M.), Texas A&M University, College Station, Texas
| |
Collapse
|
8
|
Gupta S, Gupta V. Homology modeling, structural insights and in-silico screening for selective inhibitors of mycobacterial CysE. J Biomol Struct Dyn 2020; 39:1547-1560. [PMID: 32093568 DOI: 10.1080/07391102.2020.1734089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Tuberculosis posses a major threat for health practitioners due to lengthy treatment regimen, increase in the drug-resistant strains of Mycobacterium tuberculosis (M. tb) and unavailability of drugs for its persistent form. Therefore, there is an urgent need for discovery of new and improved anti-tubercular drugs. In M. tb, the two step de novo biosynthesis of L-cysteine, an essential metabolic pathway is reported to be up-regulated in the persistent phase of the organism, involves two enzymes CysE and CysK. Although, structural insights for rational drug discovery are available for the later, not much information is known for the former. This study proposes a 3-dimensional model of M. tb CysE followed by in-silico screening of 67,030 anti-tuberculosis bioactive compounds. Subsequently, post-processing of 1000 best hits was carried out and top 200 compounds thus obtained were docked into the active site cleft of E. coli homologue as a control, but revealed unexpected results. Differences in the active site architectures and comparative analysis of molecular electrostatic potentials between the two CysEs provide molecular basis for the compounds C1, C3, C4 and C7 exhibiting preferential binding for M. tb CysE. In addition, shorter N-terminus along with positive and irregular trimeric base of M. tb CysE indicates its biological assembly as trimer. Based on mapping of residues involved in cysteine sensitivity on to the model structure of M. tb CysE, it is hypothesized that feedback inhibition of this homologue by cysteine may be affected.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sunita Gupta
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Vibha Gupta
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| |
Collapse
|
9
|
Lipničanová S, Chmelová D, Ondrejovič M, Frecer V, Miertuš S. Diversity of sialidases found in the human body - A review. Int J Biol Macromol 2020; 148:857-868. [PMID: 31945439 DOI: 10.1016/j.ijbiomac.2020.01.123] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/31/2022]
Abstract
Sialidases are enzymes essential for numerous organisms including humans. Hydrolytic sialidases (EC 3.2.1.18), trans-sialidases and anhydrosialidases (intramolecular trans-sialidases, EC 4.2.2.15) are glycoside hydrolase enzymes that cleave the glycosidic linkage and release sialic acid residues from sialyl substrates. The paper summarizes diverse sialidases present in the human body and their potential impact on development of antiviral compounds - inhibitors of viral neuraminidases. It includes a brief overview of catalytic mechanisms of action of sialidases and describes the origin of sialidases in the human body. This is followed by description of the structure and function of sialidase families with a special focus on the GH33 and GH34 families. Various effects of sialidases on human body are also briefly described. Modulation of sialidase activity may be considered a useful tool for effective treatment of various diseases. In some cases, it is desired to completely suppress the activity of sialidases by suitable inhibitors. Specific sialidase inhibitors are useful for the treatment of influenza, epilepsy, Alzheimer's disease, diabetes, different types of cancer, or heart defects. Challenges and future directions are shortly depicted in the final part of the paper.
Collapse
Affiliation(s)
- Sabina Lipničanová
- Department of Biotechnology, Faculty of Natural Sciences, University of Ss. Cyril and Methodius in Trnava, Nám. J. Herdu 2, SK-91701 Trnava, Slovakia
| | - Daniela Chmelová
- Department of Biotechnology, Faculty of Natural Sciences, University of Ss. Cyril and Methodius in Trnava, Nám. J. Herdu 2, SK-91701 Trnava, Slovakia.
| | - Miroslav Ondrejovič
- Department of Biotechnology, Faculty of Natural Sciences, University of Ss. Cyril and Methodius in Trnava, Nám. J. Herdu 2, SK-91701 Trnava, Slovakia.
| | - Vladimír Frecer
- Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, SK-83232 Bratislava, Slovakia; ICARST n.o., Jamnického 19, SK-84101, Bratislava, Slovakia.
| | - Stanislav Miertuš
- Department of Biotechnology, Faculty of Natural Sciences, University of Ss. Cyril and Methodius in Trnava, Nám. J. Herdu 2, SK-91701 Trnava, Slovakia; ICARST n.o., Jamnického 19, SK-84101, Bratislava, Slovakia.
| |
Collapse
|
10
|
Bennasroune A, Romier-Crouzet B, Blaise S, Laffargue M, Efremov RG, Martiny L, Maurice P, Duca L. Elastic fibers and elastin receptor complex: Neuraminidase-1 takes the center stage. Matrix Biol 2019; 84:57-67. [PMID: 31226402 DOI: 10.1016/j.matbio.2019.06.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/11/2019] [Accepted: 06/17/2019] [Indexed: 01/02/2023]
Abstract
Extracellular matrix (ECM) has for a long time being considered as a simple architectural support for cells. It is now clear that ECM presents a fundamental influence on cells driving their phenotype and fate. This complex network is highly specialized and the different classes of macromolecules that comprise the ECM determine its biological functions. For instance, collagens are responsible for the tensile strength of tissues, proteoglycans and glycosaminoglycans are essential for hydration and resistance to compression, and glycoproteins such as laminins facilitate cell attachment. The largest structures of the ECM are the elastic fibers found in abundance in tissues suffering high mechanical constraints such as skin, lungs or arteries. These structures present a very complex composition whose core is composed of elastin surrounded by a microfibrils mantle. Elastogenesis is a tightly regulated process involving the sialidase activity of the Neuraminidase-1 (Neu-1) sub-unit of the Elastin Receptor Complex. Interestingly, Neu-1 subunit also serves as a sensor of elastin degradation via its ability to transmit elastin-derived peptides signaling. Finally, reports showing that neuraminidase activity is able to regulate TGF-β activation raises questions about a possible role for Neu-1 in elastic fibers remodeling. In this mini review, we develop the concept of the regulation of the whole life of elastic fibers through an original scope, the key role of Neu-1 sialidase enzymatic activity.
Collapse
Affiliation(s)
- Amar Bennasroune
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | | | - Sébastien Blaise
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Muriel Laffargue
- UMR INSERM 1048 I2MC, Université Paul Sabatier, Toulouse, France
| | - Roman G Efremov
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Higher School of Economics, Myasnitskaya ul. 20, 101000 Moscow, Russia
| | - Laurent Martiny
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Pascal Maurice
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Laurent Duca
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France.
| |
Collapse
|
11
|
Guo T, Héon-Roberts R, Zou C, Zheng R, Pshezhetsky AV, Cairo CW. Selective Inhibitors of Human Neuraminidase 1 (NEU1). J Med Chem 2018; 61:11261-11279. [PMID: 30457869 DOI: 10.1021/acs.jmedchem.8b01411] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Inhibitors of human neuraminidase enzymes (NEU) are recognized as important tools for the study of the biological functions of NEU and will be potent tools for elucidating the role of these enzymes in regulating the repertoire of cellular glycans. Here we report the discovery of selective inhibitors of the human neuraminidase 1 (NEU1) and neuraminidase 2 (NEU2) enzymes with exceptional potency. A library of modified 2-deoxy-2,3-didehydro- N-acetylneuraminic acid (DANA) analogues, with variability in the C5- or C9-position, were synthesized and evaluated against four human neuraminidase isoenyzmes (NEU1-4). Hydrophobic groups with an amide linker at the C5 and C9 positions were well accommodated by NEU1, and a hexanamido group was found to give the best potency at both positions. While the C5-hexanamido-C9-hexanamido-DANA analogue did not show synergistic improvements for combined modification, an extended alkylamide at an individual position combined with a smaller group at the second gave increased potency. The best NEU1 inhibitor identified was a C5-hexanamido-C9-acetamido-DANA that had a Ki of 53 ± 5 nM and 340-fold selectivity over other isoenzymes. Additionally, we demonstrated that C5-modifications combined with a C4-guandino group provided the most potent NEU2 inhibitor reported, with a Ki of 1.3 ± 0.2 μM and 7-fold selectivity over other NEU isoenzymes.
Collapse
Affiliation(s)
- Tianlin Guo
- Alberta Glycomics Centre, Department of Chemistry , University of Alberta , Edmonton , Alberta T6G 2G2 , Canada
| | - Rachel Héon-Roberts
- Division of Medical Genetics , Sainte-Justine University Hospital Research Center, University of Montreal , Montréal , H3T 1C5 , Canada
| | - Chunxia Zou
- Alberta Glycomics Centre, Department of Chemistry , University of Alberta , Edmonton , Alberta T6G 2G2 , Canada
| | - Ruixiang Zheng
- Alberta Glycomics Centre, Department of Chemistry , University of Alberta , Edmonton , Alberta T6G 2G2 , Canada
| | - Alexey V Pshezhetsky
- Division of Medical Genetics , Sainte-Justine University Hospital Research Center, University of Montreal , Montréal , H3T 1C5 , Canada
| | - Christopher W Cairo
- Alberta Glycomics Centre, Department of Chemistry , University of Alberta , Edmonton , Alberta T6G 2G2 , Canada
| |
Collapse
|
12
|
Richards MR, Guo T, Hunter CD, Cairo CW. Molecular dynamics simulations of viral neuraminidase inhibitors with the human neuraminidase enzymes: Insights into isoenzyme selectivity. Bioorg Med Chem 2018; 26:5349-5358. [PMID: 29903413 DOI: 10.1016/j.bmc.2018.05.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/17/2018] [Accepted: 05/22/2018] [Indexed: 11/19/2022]
Abstract
Inhibitors of viral neuraminidase enzymes have been previously developed as therapeutics. Humans can express multiple forms of neuraminidase enzymes (NEU1, NEU2, NEU3, NEU4) that share a similar active site and enzymatic mechanism with their viral counterparts. Using a panel of purified human neuraminidase enzymes, we tested the inhibitory activity of 2-deoxy-2,3-dehydro-N-acetylneuraminic acid (DANA), zanamivir, oseltamivir, and peramivir against each of the human isoenzymes. We find that, with the exceptions of DANA and zanamivir, these compounds show generally poor activity against the human neuraminidase enzymes. To provide insight into the interactions of viral inhibitors with human neuraminidases, we conducted molecular dynamics simulations using homology models based on coordinates reported for NEU2. Simulations revealed that an organized water is displaced by zanamivir in binding to NEU2 and NEU3 and confirmed the critical importance of engaging the binding pocket of the C7-C9 glycerol sidechain. Our results suggest that compounds designed to target the human neuraminidases should provide more selective tools for interrogating these enzymes. Furthermore, they emphasize a need for additional structural data to enable structure-based drug design in these systems.
Collapse
Affiliation(s)
- Michele R Richards
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Tianlin Guo
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Carmanah D Hunter
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Christopher W Cairo
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada.
| |
Collapse
|
13
|
Hunter CD, Khanna N, Richards MR, Rezaei Darestani R, Zou C, Klassen JS, Cairo CW. Human Neuraminidase Isoenzymes Show Variable Activities for 9- O-Acetyl-sialoside Substrates. ACS Chem Biol 2018; 13:922-932. [PMID: 29341588 DOI: 10.1021/acschembio.7b00952] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recognition of terminal sialic acids is central to many cellular processes, and structural modification of sialic acid can disrupt these interactions. A prominent, naturally occurring, modification of sialic acid is 9- O-acetylation (9- O-Ac). Study of this modification through generation and analysis of 9- O-Ac sialosides is challenging because of the lability of the acetate group. Fundamental questions regarding the role of 9- O-Ac sialic acids remain unanswered, including what effect it may have on recognition and hydrolysis by the human neuraminidase enzymes (hNEU). To investigate the substrate activity of 9- O-acetylated sialic acids (Neu5,9Ac2), we synthesized an acetylated fluorogenic hNEU substrate 2'-(4-methylumbelliferyl)-9- O-acetyl-α-d- N-acetylneuraminic acid. Additionally, we generated a panel of octyl sialyllactosides containing modified sialic acids including variation in linkage, 9- O-acetylation, and C-5 group (Neu5Gc). Relative rates of substrate cleavage by hNEU were determined using fluorescence spectroscopy and electrospray ionization mass spectrometry. We report that 9- O-acetylation had a significant, and differential, impact on sialic acid hydrolysis by hNEU with general substrate tolerance following the trend of Neu5Ac > Neu5Gc ≫ Neu5,9Ac2 for NEU2, NEU3, and NEU4. Both NEU2 and NEU3 had remarkably reduced activity for Neu5,9Ac2 containing substrates. Other isoenzymes appeared to be more tolerant, with NEU4 even showing increased activity on Neu5,9Ac2 substrates with an aryl aglycone. The impact of these minor structural changes to sialic acid on hNEU activity was unexpected, and these results provide evidence of the substantial influence of 9- O-Ac modifications on hNEU enzyme substrate specificity. Furthermore, these findings may implicate hNEU in processes governed by 9- O-acetyltransferases and -esterases.
Collapse
Affiliation(s)
- Carmanah D. Hunter
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton Alberta T6G 2G2, Canada
| | - Neha Khanna
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton Alberta T6G 2G2, Canada
| | - Michele R. Richards
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton Alberta T6G 2G2, Canada
| | - Reza Rezaei Darestani
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton Alberta T6G 2G2, Canada
| | - Chunxia Zou
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton Alberta T6G 2G2, Canada
| | - John S. Klassen
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton Alberta T6G 2G2, Canada
| | - Christopher W. Cairo
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton Alberta T6G 2G2, Canada
| |
Collapse
|
14
|
Guo T, Dätwyler P, Demina E, Richards MR, Ge P, Zou C, Zheng R, Fougerat A, Pshezhetsky AV, Ernst B, Cairo CW. Selective Inhibitors of Human Neuraminidase 3. J Med Chem 2018; 61:1990-2008. [PMID: 29425031 DOI: 10.1021/acs.jmedchem.7b01574] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human neuraminidases (NEU) are associated with human diseases including cancer, atherosclerosis, and diabetes. To obtain small molecule inhibitors as research tools for the study of their biological functions, we designed a library of 2-deoxy-2,3-didehydro- N-acetylneuraminic acid (DANA) analogues with modifications at C4 and C9 positions. This library allowed us to discover selective inhibitors targeting the human NEU3 isoenzyme. Our most selective inhibitor for NEU3 has a Ki of 320 ± 40 nM and a 15-fold selectivity over other human neuraminidase isoenzymes. This inhibitor blocks glycolipid processing by NEU3 in vitro. To improve their pharmacokinetic properties, various esters of the best inhibitors were synthesized and evaluated. Finally, we confirmed that our best compounds exhibited selective inhibition of NEU orthologues from murine brain.
Collapse
Affiliation(s)
- Tianlin Guo
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton Alberta T6G 2G2 , Canada
| | - Philipp Dätwyler
- Department of Pharmaceutical Sciences, Pharmacenter , University of Basel , Klingelbergstrasse 50 , CH-4056 Basel , Switzerland
| | - Ekaterina Demina
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center , University of Montreal , Montréal , Quebec H3T 1C5 , Canada
| | - Michele R Richards
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton Alberta T6G 2G2 , Canada
| | - Peng Ge
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton Alberta T6G 2G2 , Canada
| | - Chunxia Zou
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton Alberta T6G 2G2 , Canada
| | - Ruixiang Zheng
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton Alberta T6G 2G2 , Canada
| | - Anne Fougerat
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center , University of Montreal , Montréal , Quebec H3T 1C5 , Canada
| | - Alexey V Pshezhetsky
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center , University of Montreal , Montréal , Quebec H3T 1C5 , Canada
| | - Beat Ernst
- Department of Pharmaceutical Sciences, Pharmacenter , University of Basel , Klingelbergstrasse 50 , CH-4056 Basel , Switzerland
| | - Christopher W Cairo
- Alberta Glycomics Centre and Department of Chemistry , University of Alberta , Edmonton Alberta T6G 2G2 , Canada
| |
Collapse
|
15
|
Haxho F, Neufeld RJ, Szewczuk MR. Neuraminidase-1: a novel therapeutic target in multistage tumorigenesis. Oncotarget 2018; 7:40860-40881. [PMID: 27029067 PMCID: PMC5130050 DOI: 10.18632/oncotarget.8396] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/18/2016] [Indexed: 12/15/2022] Open
Abstract
Several of the growth factors and their receptor tyrosine kinases (RTK) such as epidermal growth factor (EGF), platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), nerve growth factor (NGF) and insulin are promising candidate targets for cancer therapy. Indeed, tyrosine kinase inhibitors (TKI) have been developed to target these growth factors and their receptors, and have demonstrated dramatic initial responses in cancer therapy. Yet, most patients ultimately develop TKI drug resistance and relapse. It is essential in the clinical setting that the targeted therapies are to circumvent multistage tumorigenesis, including genetic mutations at the different growth factor receptors, tumor neovascularization, chemoresistance of tumors, immune-mediated tumorigenesis and the development of tissue invasion and metastasis. Here, we identify a novel receptor signaling platform linked to EGF, NGF, insulin and TOLL-like receptor (TLR) activations, all of which are known to play major roles in tumorigenesis. The importance of these findings signify an innovative and promising entirely new targeted therapy for cancer. The role of mammalian neuraminidase-1 (Neu1) in complex with matrix metalloproteinase-9 and G protein-coupled receptor tethered to RTKs and TLRs is identified as a major target in multistage tumorigenesis. Evidence exposing the link connecting growth factor-binding and immune-mediated tumorigenesis to this novel receptor-signaling paradigm will be reviewed in its current relationship to cancer.
Collapse
Affiliation(s)
- Fiona Haxho
- Departments of Biomedical and Molecular Sciences, Kingston, Ontario, Canada
| | - Ronald J Neufeld
- Department of Chemical Engineering, Queen's University, Kingston, Ontario, Canada
| | - Myron R Szewczuk
- Departments of Biomedical and Molecular Sciences, Kingston, Ontario, Canada
| |
Collapse
|
16
|
Groux-Degroote S, Rodríguez-Walker M, Dewald JH, Daniotti JL, Delannoy P. Gangliosides in Cancer Cell Signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 156:197-227. [DOI: 10.1016/bs.pmbts.2017.10.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
17
|
Paolini L, Orizio F, Busatto S, Radeghieri A, Bresciani R, Bergese P, Monti E. Exosomes Secreted by HeLa Cells Shuttle on Their Surface the Plasma Membrane-Associated Sialidase NEU3. Biochemistry 2017; 56:6401-6408. [PMID: 29039925 DOI: 10.1021/acs.biochem.7b00665] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sialidases are glycohydrolases that remove terminal sialic acid residues from oligosaccharides, glycolipids, and glycoproteins. The plasma membrane-associated sialidase NEU3 is involved in the fine-tuning of sialic acid-containing glycans directly on the cell surface and plays relevant roles in important biological phenomena such as cell differentiation, molecular recognition, and cancer transformation. Extracellular vesicles are membranous structures with a diameter of 0.03-1 μm released by cells and can be detected in blood, urine, and culture media. Among extracellular vesicles, exosomes play roles in intercellular communication and maintenance of several physiological and pathological conditions, including cancer, and could represent a useful diagnostic tool for personalized nanomedicine approaches. Using inducible expression of the murine form of NEU3 in HeLa cells, a study of the association of the enzyme with exosomes released in the culture media has been performed. Briefly, NEU3 is associated with highly purified exosomes and localizes on the external leaflet of these nanovesicles, as demonstrated by enzyme activity measurements, Western blot analysis, and dot blot analysis using specific protein markers. On the basis of these results, it is plausible that NEU3 activity on exosome glycans enhances the dynamic biological behavior of these small extracellular vesicles by modifying the negative charge and steric hindrance of their glycocalyx. The presence of NEU3 on the exosomal surface could represent a useful marker for the detection of these nanovesicles and a tool for improving our understanding of the biology of these important extracellular carriers in physiological and pathological conditions.
Collapse
Affiliation(s)
- Lucia Paolini
- Department of Molecular and Translational Medicine (DMTM), University of Brescia , 25123 Brescia, Italy
| | - Flavia Orizio
- Department of Molecular and Translational Medicine (DMTM), University of Brescia , 25123 Brescia, Italy
| | - Sara Busatto
- Department of Molecular and Translational Medicine (DMTM), University of Brescia , 25123 Brescia, Italy
| | - Annalisa Radeghieri
- Department of Molecular and Translational Medicine (DMTM), University of Brescia , 25123 Brescia, Italy
| | - Roberto Bresciani
- Department of Molecular and Translational Medicine (DMTM), University of Brescia , 25123 Brescia, Italy
| | - Paolo Bergese
- Department of Molecular and Translational Medicine (DMTM), University of Brescia , 25123 Brescia, Italy
| | - Eugenio Monti
- Department of Molecular and Translational Medicine (DMTM), University of Brescia , 25123 Brescia, Italy
| |
Collapse
|
18
|
Rodriguez-Walker M, Daniotti JL. Human Sialidase Neu3 is S-Acylated and Behaves Like an Integral Membrane Protein. Sci Rep 2017. [PMID: 28646141 PMCID: PMC5482835 DOI: 10.1038/s41598-017-04488-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Membrane-bound sialidase Neu3 is involved in the catabolism of glycoconjugates, and plays crucial roles in numerous biological processes. Since the mechanism of its association with membranes is still not completely understood, the aim of this work was to provide further information regarding this aspect. Human Neu3 was found to be associated with the plasma membrane and endomembranes, and it was not released from the lipid bilayer under conditions that typically release peripheral membrane proteins. By different experimental approaches, we demonstrated that its C-terminus is exposed to the cytosol while another portion of the protein is exposed to the extracellular space, suggesting that Neu3 possesses the features of a transmembrane protein. However, in silico analysis and homology modeling predicted that the sialidase does not contain any α-helical transmembrane segment and shares the same β-propeller fold typical of viral and bacterial sialidases. Additionally, we found that Neu3 is S-acylated. Since this post-translational modification is restricted to the cytosolic side of membranes, this finding strongly supports the idea that Neu3 may contain a cytosolic-exposed domain. Although it remains to be determined exactly how this sialidase crosses the lipid bilayer, this study provides new insights about membrane association and topology of Neu3.
Collapse
Affiliation(s)
- Macarena Rodriguez-Walker
- Centro de Investigaciones en Química Biológica de Córdoba, CIQUIBIC, CONICET and Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, Córdoba, X5000HUA, Argentina
| | - Jose L Daniotti
- Centro de Investigaciones en Química Biológica de Córdoba, CIQUIBIC, CONICET and Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, Córdoba, X5000HUA, Argentina.
| |
Collapse
|
19
|
Maurice P, Baud S, Bocharova OV, Bocharov EV, Kuznetsov AS, Kawecki C, Bocquet O, Romier B, Gorisse L, Ghirardi M, Duca L, Blaise S, Martiny L, Dauchez M, Efremov RG, Debelle L. New Insights into Molecular Organization of Human Neuraminidase-1: Transmembrane Topology and Dimerization Ability. Sci Rep 2016; 6:38363. [PMID: 27917893 PMCID: PMC5137157 DOI: 10.1038/srep38363] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 11/09/2016] [Indexed: 11/09/2022] Open
Abstract
Neuraminidase 1 (NEU1) is a lysosomal sialidase catalyzing the removal of terminal sialic acids from sialyloconjugates. A plasma membrane-bound NEU1 modulating a plethora of receptors by desialylation, has been consistently documented from the last ten years. Despite a growing interest of the scientific community to NEU1, its membrane organization is not understood and current structural and biochemical data cannot account for such membrane localization. By combining molecular biology and biochemical analyses with structural biophysics and computational approaches, we identified here two regions in human NEU1 - segments 139-159 (TM1) and 316-333 (TM2) - as potential transmembrane (TM) domains. In membrane mimicking environments, the corresponding peptides form stable α-helices and TM2 is suited for self-association. This was confirmed with full-size NEU1 by co-immunoprecipitations from membrane preparations and split-ubiquitin yeast two hybrids. The TM2 region was shown to be critical for dimerization since introduction of point mutations within TM2 leads to disruption of NEU1 dimerization and decrease of sialidase activity in membrane. In conclusion, these results bring new insights in the molecular organization of membrane-bound NEU1 and demonstrate, for the first time, the presence of two potential TM domains that may anchor NEU1 in the membrane, control its dimerization and sialidase activity.
Collapse
Affiliation(s)
- Pascal Maurice
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Stéphanie Baud
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France.,Plateau de Modélisation Moléculaire Multi-échelle, UFR Sciences Exactes et Naturelles, URCA, Reims, France
| | - Olga V Bocharova
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Eduard V Bocharov
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Andrey S Kuznetsov
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Charlotte Kawecki
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Olivier Bocquet
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Beatrice Romier
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Laetitia Gorisse
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Maxime Ghirardi
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Laurent Duca
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Sébastien Blaise
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Laurent Martiny
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Manuel Dauchez
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France.,Plateau de Modélisation Moléculaire Multi-échelle, UFR Sciences Exactes et Naturelles, URCA, Reims, France
| | - Roman G Efremov
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Higher School of Economics, Myasnitskaya ul. 20, 101000 Moscow, Russia
| | - Laurent Debelle
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| |
Collapse
|
20
|
Hyun SW, Liu A, Liu Z, Cross AS, Verceles AC, Magesh S, Kommagalla Y, Kona C, Ando H, Luzina IG, Atamas SP, Piepenbrink KH, Sundberg EJ, Guang W, Ishida H, Lillehoj EP, Goldblum SE. The NEU1-selective sialidase inhibitor, C9-butyl-amide-DANA, blocks sialidase activity and NEU1-mediated bioactivities in human lung in vitro and murine lung in vivo. Glycobiology 2016; 26:834-49. [PMID: 27226251 PMCID: PMC5884327 DOI: 10.1093/glycob/cww060] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/03/2016] [Accepted: 05/18/2016] [Indexed: 12/16/2022] Open
Abstract
Neuraminidase-1 (NEU1) is the predominant sialidase expressed in human airway epithelia and lung microvascular endothelia where it mediates multiple biological processes. We tested whether the NEU1-selective sialidase inhibitor, C9-butyl-amide-2-deoxy-2,3-dehydro-N-acetylneuraminic acid (C9-BA-DANA), inhibits one or more established NEU1-mediated bioactivities in human lung cells. We established the IC50 values of C9-BA-DANA for total sialidase activity in human airway epithelia, lung microvascular endothelia and lung fibroblasts to be 3.74 µM, 13.0 µM and 4.82 µM, respectively. In human airway epithelia, C9-BA-DANA dose-dependently inhibited flagellin-induced, NEU1-mediated mucin-1 ectodomain desialylation, adhesiveness for Pseudomonas aeruginosa and shedding. In lung microvascular endothelia, C9-BA-DANA reversed NEU1-driven restraint of cell migration into a wound and disruption of capillary-like tube formation. NEU1 and its chaperone/transport protein, protective protein/cathepsin A (PPCA), were differentially expressed in these same cells. Normalized NEU1 protein expression correlated with total sialidase activity whereas PPCA expression did not. In contrast to eukaryotic sialidases, C9-BA-DANA exerted far less inhibitory activity for three selected bacterial neuraminidases (IC50 > 800 µM). Structural modeling of the four human sialidases and three bacterial neuraminidases revealed a loop between the seventh and eighth strands of the β-propeller fold, that in NEU1, was substantially shorter than that seen in the six other enzymes. Predicted steric hindrance between this loop and C9-BA-DANA could explain its selectivity for NEU1. Finally, pretreatment of mice with C9-BA-DANA completely protected against flagellin-induced increases in lung sialidase activity. Our combined data indicate that C9-BA-DANA inhibits endogenous and ectopically expressed sialidase activity and established NEU1-mediated bioactivities in human airway epithelia, lung microvascular endothelia, and fibroblasts in vitro and murine lungs in vivo.
Collapse
Affiliation(s)
- Sang W Hyun
- Baltimore Veterans Affairs Medical Center, 10 North Greene Street, Baltimore, MD 21201, USA Department of Medicine
| | - Anguo Liu
- Baltimore Veterans Affairs Medical Center, 10 North Greene Street, Baltimore, MD 21201, USA Department of Medicine
| | - Zhenguo Liu
- Baltimore Veterans Affairs Medical Center, 10 North Greene Street, Baltimore, MD 21201, USA
| | - Alan S Cross
- Department of Medicine Center for Vaccine Development, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA
| | | | - Sadagopan Magesh
- Department of Applied Bio-organic Chemistry, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Yadagiri Kommagalla
- Department of Applied Bio-organic Chemistry, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Chandrababunaidu Kona
- Department of Applied Bio-organic Chemistry, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-honmachi, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiromune Ando
- Department of Applied Bio-organic Chemistry, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-honmachi, Sakyo-ku, Kyoto 606-8501, Japan
| | - Irina G Luzina
- Baltimore Veterans Affairs Medical Center, 10 North Greene Street, Baltimore, MD 21201, USA Department of Medicine
| | - Sergei P Atamas
- Baltimore Veterans Affairs Medical Center, 10 North Greene Street, Baltimore, MD 21201, USA Department of Medicine Department of Microbology and Immunology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA
| | - Kurt H Piepenbrink
- Department of Medicine Institute of Human Virology, University of Maryland School of Medicine, 725 West Lombard St, Baltimore, MD 21201, USA
| | - Eric J Sundberg
- Department of Medicine Department of Microbology and Immunology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA Institute of Human Virology, University of Maryland School of Medicine, 725 West Lombard St, Baltimore, MD 21201, USA
| | - Wei Guang
- Department of Pediatrics, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA
| | - Hideharu Ishida
- Department of Applied Bio-organic Chemistry, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Erik P Lillehoj
- Department of Pediatrics, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA
| | - Simeon E Goldblum
- Baltimore Veterans Affairs Medical Center, 10 North Greene Street, Baltimore, MD 21201, USA Center for Vaccine Development, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201, USA
| |
Collapse
|
21
|
Sriwilaijaroen N, Magesh S, Imamura A, Ando H, Ishida H, Sakai M, Ishitsubo E, Hori T, Moriya S, Ishikawa T, Kuwata K, Odagiri T, Tashiro M, Hiramatsu H, Tsukamoto K, Miyagi T, Tokiwa H, Kiso M, Suzuki Y. A Novel Potent and Highly Specific Inhibitor against Influenza Viral N1-N9 Neuraminidases: Insight into Neuraminidase-Inhibitor Interactions. J Med Chem 2016; 59:4563-77. [PMID: 27095056 DOI: 10.1021/acs.jmedchem.5b01863] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
People throughout the world continue to be at risk for death from influenza A virus, which is always creating a new variant. Here we present a new effective and specific anti-influenza viral neuraminidase (viNA) inhibitor, 9-cyclopropylcarbonylamino-4-guanidino-Neu5Ac2en (cPro-GUN). Like zanamivir, it is highly effective against N1-N9 avian and N1-N2 human viNAs, including H274Y oseltamivir-resistant N1 viNA, due to its C-6 portion still being anchored in the active site, different from the disruption of oseltamivir's C-6 anchoring by H274Y mutation. Unlike zanamivir, no sialidase inhibitory activity has been observed for cPro-GUN against huNeu1-huNeu4 enzymes. Broad efficacy of cPro-GUN against avian and human influenza viruses in cell cultures comparable to its sialidase inhibitory activities makes cPro-GUN ideal for further development for safe therapeutic or prophylactic use against both seasonal and pandemic influenza.
Collapse
Affiliation(s)
- Nongluk Sriwilaijaroen
- Department of Preclinical Sciences, Faculty of Medicine, Thammasat University , Pathumthani 12120, Thailand.,Health Science Hills, College of Life and Health Sciences, Chubu University , Aichi 487-8501, Japan
| | | | | | - Hiromune Ando
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University , Kyoto 606-8501, Japan
| | | | | | | | | | - Setsuko Moriya
- Division of Cancer Glycosylation Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University , Sendai 981-8558, Japan
| | | | | | - Takato Odagiri
- Influenza Virus Research Center, National Institute of Infectious Diseases , Tokyo 208-0011, Japan
| | - Masato Tashiro
- Influenza Virus Research Center, National Institute of Infectious Diseases , Tokyo 208-0011, Japan
| | - Hiroaki Hiramatsu
- Health Science Hills, College of Life and Health Sciences, Chubu University , Aichi 487-8501, Japan
| | - Kenji Tsukamoto
- Research Team for Zoonotic Diseases, National Institute of Animal Health , Ibaraki 305-0856, Japan
| | - Taeko Miyagi
- Division of Cancer Glycosylation Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University , Sendai 981-8558, Japan
| | | | - Makoto Kiso
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University , Kyoto 606-8501, Japan
| | - Yasuo Suzuki
- Health Science Hills, College of Life and Health Sciences, Chubu University , Aichi 487-8501, Japan
| |
Collapse
|
22
|
Rahman MM, Hirokawa T, Tsuji D, Tsukimoto J, Hitaoka S, Yoshida T, Chuman H, Itoh K. Novel pH-dependent regulation of human cytosolic sialidase 2 (NEU2) activities by siastatin B and structural prediction of NEU2/siastatin B complex. Biochem Biophys Rep 2015; 4:234-242. [PMID: 29124209 PMCID: PMC5669518 DOI: 10.1016/j.bbrep.2015.09.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 09/20/2015] [Accepted: 09/22/2015] [Indexed: 11/18/2022] Open
Abstract
Human cytosolic sialidase (Neuraminidase 2, NEU2) catalyzes the removal of terminal sialic acid residues from glycoconjugates. The effect of siastatin B, known as a sialidase inhibitor, has not been evaluated toward human NEU2 yet. We studied the regulation of NEU2 activity by siastatin B in vitro and predicted the interaction in silico. Inhibitory and stabilizing effects of siastatin B were analyzed in comparison with DANA (2-deoxy-2,3-dehydro-N-acetylneuraminic acid) toward 4-umbelliferyl N-acetylneuraminic acid (4-MU-NANA)- and α2,3-sialyllactose-degrading activities of recombinant NEU2 produced by E. coli GST-fusion gene expression. Siastatin B exhibited to have higher competitive inhibitory activity toward NEU2 than DANA at pH 4.0. We also revealed the stabilizing effect of siastatin B toward NEU2 activity at acidic pH. Docking model was constructed on the basis of the crystal structure of NEU2/DANA complex (PDB code: 1VCU). Molecular docking predicted that electrostatic neutralization of E111 and E218 residues of the active pocket should not prevent siastatin B from binding at pH 4.0. The imino group (1NH) of siastatin B can also interact with D46, neutralized at pH 4.0. Siastatin B was suggested to have higher affinity to the active pocket of NEU2 than DANA, although it has no C7-9 fragment corresponding to that of DANA. We demonstrated here the pH-dependent affinity of siastatin B toward NEU2 to exhibit potent inhibitory and stabilizing activities. Molecular interaction between siastatin B and NEU2 will be utilized to develop specific inhibitors and stabilizers (chemical chaperones) not only for NEU2 but also the other human sialidases, including NEU1, NEU3 and NEU4, based on homology modeling.
Collapse
Affiliation(s)
- M. Motiur Rahman
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Takatsugu Hirokawa
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), 2-42 Aomi, Koto-ku, Tokyo 135-0064, Japan
| | - Daisuke Tsuji
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Jun Tsukimoto
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Seiji Hitaoka
- Department of Theoretical Chemistry for Drug Discovery, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Tatsusada Yoshida
- Department of Theoretical Chemistry for Drug Discovery, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Hiroshi Chuman
- Department of Theoretical Chemistry for Drug Discovery, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Kohji Itoh
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
- Corresponding author.
| |
Collapse
|
23
|
Zamora CY, Ryan MJ, d'Alarcao M, Kumar K. Sialidases as regulators of bioengineered cellular surfaces. Glycobiology 2015; 25:784-91. [PMID: 25795684 DOI: 10.1093/glycob/cwv019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 03/11/2015] [Indexed: 12/13/2022] Open
Abstract
Human sialidases (NEUs) catalyze the removal of N-acetyl neuraminic acids from the glycome of the cell and regulate a diverse repertoire of nominal cellular functions, such as cell signaling and adhesion. A greater understanding of their substrate permissivity is of interest in order to discern their physiological functions in disease states and in the design of specific and effective small molecule inhibitors. Towards this, we have synthesized soluble fluorogenic reporters of mammalian sialidase activity bearing unnatural sialic acids commonly incorporated into the cellular glycocalyx via metabolic glycoengineering. We found cell-surface sialidases in Jurkat capable of cleaving unnatural sialic acids with differential activities toward a variety of R groups on neuraminic acid. In addition, we observed modulated structure-activity relationships when cell-surface sialidases were presented glycans with unnatural bulky, hydrophobic or fluorinated moieties incorporated directly via glycoengineering. Our results confirm the importance of cell-surface sialidases in glycoengineering incorporation data. We demonstrate the flexibility of human NEUs toward derivatized sugars and highlight the importance of native glycan presentation to sialidase binding and activity. These results stand to inform not only metabolic glycoengineering efforts but also inhibitor design.
Collapse
Affiliation(s)
| | - Matthew J Ryan
- Department of Chemistry, Tufts University, Medford, MA, USA
| | - Marc d'Alarcao
- Department of Chemistry, San José State University, San José, CA, USA
| | - Krishna Kumar
- Department of Chemistry, Tufts University, Medford, MA, USA Cancer Center, Tufts Medical Center Boston, Boston, MA, USA
| |
Collapse
|
24
|
Shiozaki K, Takahashi K, Hosono M, Yamaguchi K, Hata K, Shiozaki M, Bassi R, Prinetti A, Sonnino S, Nitta K, Miyagi T. Phosphatidic acid-mediated activation and translocation to the cell surface of sialidase NEU3, promoting signaling for cell migration. FASEB J 2015; 29:2099-111. [PMID: 25678627 DOI: 10.1096/fj.14-262543] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 01/11/2015] [Indexed: 11/11/2022]
Abstract
The plasma membrane-associated sialidase NEU3 plays crucial roles in regulation of transmembrane signaling, and its aberrant up-regulation in various cancers contributes to malignancy. However, it remains uncertain how NEU3 is naturally activated and locates to plasma membranes, because of its Triton X-100 requirement for the sialidase activity in vitro and its often changing subcellular location. Among phospholipids examined, we demonstrate that phosphatidic acid (PA) elevates its sialidase activity 4 to 5 times at 50 μM in vitro at neutral pH and promotes translocation to the cell surface and cell migration through Ras-signaling in HeLa and COS-1 cells. NEU3 was found to interact selectively with PA as assessed by phospholipid array, liposome coprecipitation, and ELISA assays and to colocalize with phospholipase D (PLD) 1 in response to epidermal growth factor (EGF) or serum stimulation. Studies using tagged NEU3 fragments with point mutations identified PA- and calmodulin (CaM)-binding sites around the N terminus and confirmed its participation in translocation and catalytic activity. EGF induced PLD1 activation concomitantly with enhanced NEU3 translocation to the cell surface, as assessed by confocal microscopy. These results suggest that interactions of NEU3 with PA produced by PLD1 are important for regulation of transmembrane signaling, this aberrant acceleration probably promoting malignancy in cancers.
Collapse
Affiliation(s)
- Kazuhiro Shiozaki
- *Faculty of Fisheries and The United Graduate School of Agricultural Science, Kagoshima University, Kagoshima, Kagoshima, Japan; Division of Cancer Glycosylation Research and Division of Cell Recognition Study, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan; and Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy
| | - Kohta Takahashi
- *Faculty of Fisheries and The United Graduate School of Agricultural Science, Kagoshima University, Kagoshima, Kagoshima, Japan; Division of Cancer Glycosylation Research and Division of Cell Recognition Study, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan; and Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy
| | - Masahiro Hosono
- *Faculty of Fisheries and The United Graduate School of Agricultural Science, Kagoshima University, Kagoshima, Kagoshima, Japan; Division of Cancer Glycosylation Research and Division of Cell Recognition Study, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan; and Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy
| | - Kazunori Yamaguchi
- *Faculty of Fisheries and The United Graduate School of Agricultural Science, Kagoshima University, Kagoshima, Kagoshima, Japan; Division of Cancer Glycosylation Research and Division of Cell Recognition Study, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan; and Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy
| | - Keiko Hata
- *Faculty of Fisheries and The United Graduate School of Agricultural Science, Kagoshima University, Kagoshima, Kagoshima, Japan; Division of Cancer Glycosylation Research and Division of Cell Recognition Study, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan; and Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy
| | - Momo Shiozaki
- *Faculty of Fisheries and The United Graduate School of Agricultural Science, Kagoshima University, Kagoshima, Kagoshima, Japan; Division of Cancer Glycosylation Research and Division of Cell Recognition Study, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan; and Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy
| | - Rosaria Bassi
- *Faculty of Fisheries and The United Graduate School of Agricultural Science, Kagoshima University, Kagoshima, Kagoshima, Japan; Division of Cancer Glycosylation Research and Division of Cell Recognition Study, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan; and Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy
| | - Alessandro Prinetti
- *Faculty of Fisheries and The United Graduate School of Agricultural Science, Kagoshima University, Kagoshima, Kagoshima, Japan; Division of Cancer Glycosylation Research and Division of Cell Recognition Study, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan; and Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy
| | - Sandro Sonnino
- *Faculty of Fisheries and The United Graduate School of Agricultural Science, Kagoshima University, Kagoshima, Kagoshima, Japan; Division of Cancer Glycosylation Research and Division of Cell Recognition Study, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan; and Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy
| | - Kazuo Nitta
- *Faculty of Fisheries and The United Graduate School of Agricultural Science, Kagoshima University, Kagoshima, Kagoshima, Japan; Division of Cancer Glycosylation Research and Division of Cell Recognition Study, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan; and Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy
| | - Taeko Miyagi
- *Faculty of Fisheries and The United Graduate School of Agricultural Science, Kagoshima University, Kagoshima, Kagoshima, Japan; Division of Cancer Glycosylation Research and Division of Cell Recognition Study, Tohoku Pharmaceutical University, Sendai, Miyagi, Japan; Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan; and Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy
| |
Collapse
|
25
|
Tomin A, Dumych T, Tolstyak Y, Kril I, Mahorivska I, Bila E, Stoika R, Herrmann M, Kit Y, Bilyy R. Desialylation of dying cells with catalytically active antibodies possessing sialidase activity facilitate their clearance by human macrophages. Clin Exp Immunol 2015; 179:17-23. [PMID: 24580640 DOI: 10.1111/cei.12312] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2014] [Indexed: 12/26/2022] Open
Abstract
Recently we reported the first known incidence of antibodies possessing catalytic sialidase activity (sialidase abzymes) in the serum of patients with multiple myeloma and systemic lupus erythematosus (SLE). These antibodies desialylate biomolecules, such as glycoproteins, gangliosides and red blood cells. Desialylation of dying cells was demonstrated to facilitate apoptotic cell clearance. In this study we assessed the possibility to facilitate dying cell clearance with the use of F(ab)2 fragments of sialidase abzymes. Two sources of sialidase abzymes were used: (i) those isolated from sera of patients with SLE after preliminary screening of a cohort of patients for sialidase activity; and (ii) by creating an induced sialidase abzyme through immunization of a rabbit with synthetic hapten consisting of a non-hydrolysable analogue of sialidase reaction conjugated with bovine serum albumin (BSA) or keyhole limpet haemocyanin (KLH). Antibodies were purified by ammonium sulphate precipitation, protein-G affinity chromatography and size exclusion-high performance liquid chromatography (HPLC-SEC). Effect of desialylation on efferocytosis was studied using human polymorphonuclear leucocytes (PMN), both viable and aged, as prey, and human monocyte-derived macrophages (MoMa). Treatment of apoptotic and viable prey with both disease-associated (purified from blood serum of SLE patients) and immunization-induced (obtained by immunization of rabbits) sialidase abzymes, its F(ab)2 fragment and bacterial neuraminidase (as positive control) have significantly enhanced the clearance of prey by macrophages. We conclude that sialidase abzyme can serve as a protective agent in autoimmune patients and that artificial abzymes may be of potential therapeutic value.
Collapse
Affiliation(s)
- A Tomin
- Institute of Cell Biology, National Academy of Sciences of Ukraine, Lviv, Ukraine
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Kwak JE, Son MY, Son YS, Son MJ, Cho YS. Biochemical and molecular characterization of novel mutations in GLB1 and NEU1 in patient cells with lysosomal storage disorders. Biochem Biophys Res Commun 2015; 457:554-60. [PMID: 25600812 DOI: 10.1016/j.bbrc.2015.01.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/08/2015] [Indexed: 12/22/2022]
Abstract
Lysosomes are cytoplasmic compartments that contain many acid hydrolases and play critical roles in the metabolism of a wide range of macromolecules. Deficiencies in lysosomal enzyme activities cause genetic diseases, called lysosomal storage disorders (LSDs). Many mutations have been identified in the genes responsible for LSDs, and the identification of mutations is required for the accurate molecular diagnoses. Here, we analyzed cell lines that were derived from two different LSDs, GM1 gangliosidosis and sialidosis. GM1 gangliosidosis is caused by mutations in the GLB1 gene that encodes β-galactosidase. A lack of β-galactosidase activity leads to the massive accumulation of GM1 ganglioside, which results in neurodegenerative pathology. Mutations in the NEU1 gene that encodes lysosomal sialidase cause sialidosis. Insufficient activity of lysosomal sialidase progressively increases the accumulation of sialylated molecules, and various clinical symptoms, including mental retardation, appear. We sequenced the entire coding regions of GLB1 and NEU1 in GM1 gangliosidosis and sialidosis patient cells, respectively. We found the novel mutations p.E186A in GLB1 and p.R347Q in NEU1, as well as many other mutations that have been previously reported. We also demonstrated that patient cells containing the novel mutations showed the molecular phenotypes of the corresponding disease. Further structural analysis suggested that these novel mutation sites are highly conserved and important for enzyme activity.
Collapse
Affiliation(s)
- Jae Eun Kwak
- Stem Cell Research Center, KRIBB, 111 Gwahangno, Yuseong-gu, Daejeon 305-806, Republic of Korea
| | - Mi-Young Son
- Stem Cell Research Center, KRIBB, 111 Gwahangno, Yuseong-gu, Daejeon 305-806, Republic of Korea; Department of Functional Genomics, University of Science & Technology, 217 Gajungro, Yuseong-gu, Daejeon 305-350, Republic of Korea
| | - Ye Seul Son
- Stem Cell Research Center, KRIBB, 111 Gwahangno, Yuseong-gu, Daejeon 305-806, Republic of Korea
| | - Myung Jin Son
- Stem Cell Research Center, KRIBB, 111 Gwahangno, Yuseong-gu, Daejeon 305-806, Republic of Korea; Department of Functional Genomics, University of Science & Technology, 217 Gajungro, Yuseong-gu, Daejeon 305-350, Republic of Korea
| | - Yee Sook Cho
- Stem Cell Research Center, KRIBB, 111 Gwahangno, Yuseong-gu, Daejeon 305-806, Republic of Korea; Department of Functional Genomics, University of Science & Technology, 217 Gajungro, Yuseong-gu, Daejeon 305-350, Republic of Korea.
| |
Collapse
|
27
|
Albohy A, Richards MR, Cairo CW. Mapping substrate interactions of the human membrane-associated neuraminidase, NEU3, using STD NMR. Glycobiology 2014; 25:284-93. [DOI: 10.1093/glycob/cwu109] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
28
|
Abstract
A review of known small molecule inhibitors and substrates of the human neuraminidase enzymes.
Collapse
Affiliation(s)
- Christopher W. Cairo
- Alberta Glycomics Centre
- Department of Chemistry
- University of Alberta
- Edmonton Alberta
- Canada
| |
Collapse
|
29
|
Zhang Y, Albohy A, Zou Y, Smutova V, Pshezhetsky AV, Cairo CW. Identification of selective inhibitors for human neuraminidase isoenzymes using C4,C7-modified 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (DANA) analogues. J Med Chem 2013; 56:2948-58. [PMID: 23530623 DOI: 10.1021/jm301892f] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the past two decades, human neuraminidases (human sialidases, hNEUs) have been found to be involved in numerous pathways in biology. The development of selective and potent inhibitors of these enzymes will provide critical tools for glycobiology, help to avoid undesired side effects of antivirals, and may reveal new small-molecule therapeutic targets for human cancers. However, because of the high active site homology of the hNEU isoenzymes, little progress in the design and synthesis of selective inhibitors has been realized. Guided by our previous studies of human NEU3 inhibitors, we designed a series of C4,C7-modified analogues of 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (DANA) and tested them against the full panel of hNEU isoenzymes (NEU1, NEU2, NEU3, NEU4). We identified inhibitors with up to 38-fold selectivity for NEU3 and 12-fold selectivity for NEU2 over all other isoenzymes. We also identified compounds that targeted NEU2 and NEU3 with similar potency.
Collapse
Affiliation(s)
- Yi Zhang
- Alberta Glycomics Center, Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | | | | | | | | | | |
Collapse
|
30
|
Rahman MM, Kitao S, Tsuji D, Suzuki K, Sakamoto JI, Matsuoka K, Matsuzawa F, Aikawa SI, Itoh K. Inhibitory effects and specificity of synthetic sialyldendrimers toward recombinant human cytosolic sialidase 2 (NEU2). Glycobiology 2013; 23:495-504. [DOI: 10.1093/glycob/cws221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
31
|
O'Leary EM, Igdoura SA. The therapeutic potential of pharmacological chaperones and proteosomal inhibitors, Celastrol and MG132 in the treatment of sialidosis. Mol Genet Metab 2012; 107:173-85. [PMID: 22898113 DOI: 10.1016/j.ymgme.2012.07.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 07/12/2012] [Accepted: 07/12/2012] [Indexed: 10/28/2022]
Abstract
Sialidosis is an autosomal recessive disorder caused by a dysfunctional Sialidase enzyme. Categorised into two phenotypes, Sialidosis type I and II, Sialidosis is a highly heterogeneous disorder with varying ages of onset and pathologies. Currently, there is no viable therapy for the treatment of Sialidosis patients. At the molecular level, cells from Sialidosis patients with compound heterozygous mutations show improper enzyme folding, loss of Sialidase enzyme activity and subsequent accumulation of sialylconjugates within lysosomes. One promising treatment option is the use of small pharmacological molecules to increase the enzymatic activities of mutant proteins. In this study, we examined the efficacy of the immuno-suppressant (Celastrol) as well as a proteosomal inhibitor (MG132) to rescue mutant enzymes with altered conformation. Our results reveal that MG132 enhances enzyme activity and its localisation in cells expressing defective Sialidase. We also found that MG132 reduces accumulation of ganglioside products, GT1b, GD3, and GM3 in pre-loaded Sialidosis cells. Alternatively, Celastrol appears to reduce Sialidase expression and activity revealing a potentially novel effect of Celastrol on Sialidase. Interestingly, the combination of Celastrol and MG132 appears to amplify the beneficial impact of MG132 on both the endogenous and recombinant expression of defective Sialidase. This study explores a novel biological criteria to assess the efficacy of small molecules through accumulation analysis and points to a potential therapeutic strategy for the treatment of Sialidosis.
Collapse
Affiliation(s)
- Erin M O'Leary
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
32
|
Mozzi A, Mazzacuva P, Zampella G, Forcella ME, Fusi PA, Monti E. Molecular insight into substrate recognition by human cytosolic sialidase NEU2. Proteins 2012; 80:1123-32. [PMID: 22228546 DOI: 10.1002/prot.24013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 11/29/2011] [Accepted: 12/01/2011] [Indexed: 12/18/2022]
Abstract
Sialidases or neuramidases are glycoside hydrolases removing terminal sialic acid residues from sialo-glycoproteins and sialo-glycolipids. Viral neuraminidases (NAs) have been extensively characterized and represent an excellent target for antiviral therapy through the synthesis of a series of competitive inhibitors that block the release of newly formed viral particles from infected cells. The human cytosolic sialidase NEU2 is the only mammalian enzyme structurally characterized and represents a valuable model to study the specificity of novel NA inhibitory drugs. Moreover, the availability of NEU2 3D structure represents a pivotal step toward the characterization of the molecular basis of natural substrates recognition by the enzyme. In this perspective, we have carried out a study of molecular docking of NEU2 active site using natural substrates of increasing complexity. Moreover, selective mutations of the residues putatively involved into substrate(s) interaction/recognition have been performed, and the resulting mutant enzymes have been preliminary tested for their catalytic activity and substrate specificity. We found that Q270 is involved in the binding of the disaccharide α(2,3) sialyl-galactose, whereas K45 and Q112 bind the distal glucose of the trisaccharide α(2,3) sialyl-lactose, corresponding to the oligosaccharide moiety of GM3 ganglioside. In addition, E218, beside D46, is proved to be a key catalytic residue, being, together with Y334, the second member of the nucleophile pair required for the catalysis. Overall, our results point out the existence of a dynamic network of interactions that are possibly involved in the recognition of the glycans bearing sialic acid.
Collapse
Affiliation(s)
- Alessandra Mozzi
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Milano, Italy
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
The removal of sialic acids, catalyzed by sialidase, is the initial step in degradation of oligosaccharides, glycoproteins, and glycolipids. The catalytic reaction may greatly influence biological processes through changing the conformation of glycoproteins and create or mask binding sites of functional molecules. Recent progress in sialidase research has clarified that mammalian sialidases indeed contribute to the regulation of various cellular functions as well as lysosomal catabolism, unlike the sialidases of microbial origin that probably play roles limited to nutrition and pathogenesis. However, the mammalian enzymes contain consensus sequences in the six-blade β-propeller structural organization typical of microbial sialidases, despite the low degree of similarity to the amino acid sequences of the microbial enzymes. The present review briefly summarizes structural and functional features of mammalian sialidases.
Collapse
Affiliation(s)
- Eugenio Monti
- Faculty of Medicine, Department of Biomedical Sciences and Biotechnology, University of Brescia, Brescia, Viale Europa 11, 25123, Brescia, Italy
| | | |
Collapse
|
34
|
Wickramasinghe S, Medrano JF. Primer on genes encoding enzymes in sialic acid metabolism in mammals. Biochimie 2011; 93:1641-6. [PMID: 21689720 DOI: 10.1016/j.biochi.2011.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Accepted: 06/03/2011] [Indexed: 11/18/2022]
Abstract
Sialic acid, a nine-carbon sugar acid usually is present in the non-reducing terminal position of free oligosaccharides and glycoconjugates. Sialylated conjugates in mammals perform important roles in cellular recognition, signaling, host-pathogen interaction and neuronal development. Metabolism of sialylated conjugates involves a complex pathway consisting of enzymes distributed among the different compartments in the cell. These enzymes are encoded by 32 genes diversely distributed throughout the mammalian genome. Genetic variants in some of these genes are associated with embryonic lethality and abnormal phenotypes in mice and neuromuscular diseases, carcinomas and immune-mediated diseases in humans. In humans, the CMP-NeuAc-hydroxylase (CMAH) enzyme is inactivated due to a deletion mutation in the encoded enzyme. This lack of Neu5Gc phenotype makes humans unique among mammals. This review focuses on genes encoding enzymes in sialic acid metabolism pathways in mammalian cells with special emphasis on the human, mouse and cow.
Collapse
Affiliation(s)
- Saumya Wickramasinghe
- Department of Animal Science, University of California-Davis, One Shields Ave., Davis, CA 95616-8521, USA
| | | |
Collapse
|
35
|
Coutinho MF, Lacerda L, Macedo-Ribeiro S, Baptista E, Ribeiro H, Prata MJ, Alves S. Lysosomal multienzymatic complex-related diseases: a genetic study among Portuguese patients. Clin Genet 2011; 81:379-93. [PMID: 21214877 DOI: 10.1111/j.1399-0004.2011.01625.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The functional activity of lysosomal enzymes sialidase, β-galactosidase and N-acetylaminogalacto-6-sulfate-sulfatase in the cell depends on their association in a multienzyme complex with cathepsin A. Mutations in any of the components of this complex result in functional deficiency thereby causing severe lysosomal storage disorders. Here, we report the molecular defects underlying sialidosis (mutations in sialidase; gene NEU1), galactosialidosis (mutations in cathepsin A; gene PPGB) and GM1 gangliosidosis (mutations in β-galactosidase; gene GLB1) in Portuguese patients. We performed molecular studies of the PPGB, NEU1 and GLB1 genes in biochemically diagnosed Portuguese patients. Gene expression was determined and the effect of each mutation predicted at protein levels. In the NEU1 gene, we found three novel missense mutations (p.P200L, p.D234N and p.Q282H) and one nonsense mutation (p.R341X). In the PPGB gene, we identified two missense mutations, one novel (p.G86V) and one already described (p.V104M), as well as two new deletions (c.230delC and c.991-992delT) that give rise to non-functional proteins. We also present the first molecular evidence of a causal missense mutation localized to the cathepsin A active site. Finally, in the GLB1 gene, we found six different mutations, all of them previously described (p.R59H, p.R201H, p.H281Y, p.W527X, c.1572-1577InsG and c.845-846delC). Seven novel mutations are reported here, contributing to our knowledge of the mutational spectrum of these diseases and to a better understanding of the genetics of the lysosomal multienzymatic complex. The results of this study will allow carrier detection in affected families and prenatal molecular diagnosis, leading to the improvement of genetic counseling.
Collapse
Affiliation(s)
- M F Coutinho
- Centro de Genética Médica Doutor Jacinto de Magalhães, INSA, I.P., Praça Pedro Nunes 88, Porto, Portugal.
| | | | | | | | | | | | | |
Collapse
|
36
|
Li Y, Cao H, Yu H, Chen Y, Lau K, Qu J, Thon V, Sugiarto G, Chen X. Identifying selective inhibitors against the human cytosolic sialidase NEU2 by substrate specificity studies. MOLECULAR BIOSYSTEMS 2011; 7:1060-72. [PMID: 21206954 DOI: 10.1039/c0mb00244e] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aberrant expression of human sialidases has been shown to associate with various pathological conditions. Despite the effort in the sialidase inhibitor design, less attention has been paid to designing specific inhibitors against human sialidases and characterizing the substrate specificity of different sialidases regarding diverse terminal sialic acid forms and sialyl linkages. This is mainly due to the lack of sialoside probes and efficient screening methods, as well as limited access to human sialidases. A low cellular expression level of the human sialidase NEU2 hampers its functional and inhibitory studies. Here we report the successful cloning and expression of the human sialidase NEU2 in E. coli. About 11 mg of soluble active NEU2 was routinely obtained from 1 L of E. coli cell culture. Substrate specificity studies of the recombinant human NEU2 using twenty p-nitrophenol (pNP)-tagged α2-3- or α2-6-linked sialyl galactosides containing different terminal sialic acid forms including common N-acetylneuraminic acid (Neu5Ac), non-human N-glycolylneuraminic acid (Neu5Gc), 2-keto-3-deoxy-D-glycero-D-galacto-nonulosonic acid (Kdn), or their C5-derivatives in a microtiter plate-based high-throughput colorimetric assay identified a unique structural feature specifically recognized by the human NEU2 but not two bacterial sialidases. The results obtained from substrate specificity studies were used to guide the design of a sialidase inhibitor that was selective against human NEU2. The selectivity of the inhibitor was revealed by the comparison of sialidase crystal structures and inhibitor docking studies.
Collapse
Affiliation(s)
- Yanhong Li
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA 95616, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Glycosphingolipids and Kidney Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 721:121-38. [PMID: 21910086 DOI: 10.1007/978-1-4614-0650-1_8] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
38
|
Evaluation of a Set of C9 N-acyl Neu5Ac2en Mimetics as Viral Sialidase Selective Inhibitors. INTERNATIONAL JOURNAL OF MEDICINAL CHEMISTRY 2010; 2011:539245. [PMID: 27525119 PMCID: PMC4971745 DOI: 10.1155/2011/539245] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2010] [Revised: 11/16/2010] [Accepted: 11/16/2010] [Indexed: 11/17/2022]
Abstract
Identification of selective influenza viral sialidase inhibitors is highly desirable in order to minimize or avoid the adverse effects due to the possible inhibition of endogenous human sialidases. We recently reported the evaluation of C9 N-acyl Neu5Ac2en mimetics as probes for human sialidases. Herein, we describe the in vitro activity of the same set of C9 N-acyl Neu5Ac2en mimetics against sialidases expressed by influenza virus A/PR/8/34 (H1N1), A/Memphis/1/72 (H3N2), and A/Duck/313/78 (H5N3) strains. Compound 8 is identified as a promising starting point for the development of viral sialidase selective inhibitors. Multiple sequence alignment and molecular docking techniques are also performed to explore the plausible interaction of compound 8 with viral sialidases.
Collapse
|
39
|
Zou Y, Albohy A, Sandbhor M, Cairo CW. Inhibition of human neuraminidase 3 (NEU3) by C9-triazole derivatives of 2,3-didehydro-N-acetyl-neuraminic acid. Bioorg Med Chem Lett 2010; 20:7529-33. [PMID: 21036040 DOI: 10.1016/j.bmcl.2010.09.111] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 09/19/2010] [Accepted: 09/22/2010] [Indexed: 12/13/2022]
Abstract
We report the synthesis of a series of C9 and N5Ac modified analogs of 2,3-didehydro-N-acetyl-neuraminic acid (DANA) and their inhibitory potency for the human neuraminidase 3 (NEU3) enzyme. We were able to generate a small library of compounds through the synthesis of azide derivatives of DANA, followed by Cu-catalyzed azide-alkyne cycloaddition (CuAAC) to generate triazole-containing inhibitors. Our results suggest that NEU3 can tolerate large hydrophobic groups at the C9 position; however, none of the derivatives made at the N5Ac side-chain were active. We identify three new inhibitors that have comparable potency to the best reported inhibitors of the enzyme.
Collapse
Affiliation(s)
- Yao Zou
- Alberta Ingenuity Centre for Carbohydrate Science, Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | |
Collapse
|
40
|
Albohy A, Li MD, Zheng RB, Zou C, Cairo CW. Insight into substrate recognition and catalysis by the human neuraminidase 3 (NEU3) through molecular modeling and site-directed mutagenesis. Glycobiology 2010; 20:1127-38. [DOI: 10.1093/glycob/cwq077] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
|
41
|
Levy R, Edelman M, Sobolev V. Prediction of 3D metal binding sites from translated gene sequences based on remote-homology templates. Proteins 2010; 76:365-74. [PMID: 19173310 DOI: 10.1002/prot.22352] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Database-scale analysis was performed to determine whether structural models, based on remote homologues, are effective in predicting 3D transition metal binding sites in proteins directly from translated gene sequences. The extent by which side chain modeling alone reduces sensitivity and selectivity is shown to be <10%. Surprisingly, selectivity was not dependent on the level of sequence homology between template and target, or on the presence of a metal ion in the structural template. Applying a modification of the CHED algorithm (Babor et al., Proteins 2008;70:208-217) and machine learning filters, a selectivity of approximately 90% was achieved for protein sequences using unrelated structural templates over a sequence identity range of 18-100%. Below approximately 18% identity, the number of analyzable target-template pairs and predictability of metal binding sites falls off sharply. A full third of structural templates were found to have target partners only in the remote homology range of 18-30%. In this range, nonmetal-binding templates are calculated to be the majority and serve to predict with 50% sensitivity at the geometric level. Overall, sensitivity at the geometric level for targets having templates in the 18-30% sequence identity range is 73%, with an average of one false positive site per true site. Protein sequences described as "unknown" in the UniProt database and composed largely of unidentified genome project sequences were studied and metal binding sites predicted. A web server for prediction of metal binding sites from protein sequence is provided.
Collapse
Affiliation(s)
- Ronen Levy
- Department of Plant Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | | |
Collapse
|
42
|
Tiwari M, Lee JK. Molecular modeling studies of L-arabinitol 4-dehydrogenase of Hypocrea jecorina: its binding interactions with substrate and cofactor. J Mol Graph Model 2010; 28:707-13. [PMID: 20171913 DOI: 10.1016/j.jmgm.2010.01.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Accepted: 01/09/2010] [Indexed: 10/20/2022]
Abstract
L-arabinitol 4-dehydrogenase (LAD1; EC 1.1.1.12) is an enzyme in the L-arabinose catabolic pathway of fungi that catalyzes the conversion of L-arabinitol into L-xylulose. The primary objective of this work is to identify the catalytic and coenzyme binding domains of LAD1 from Hypocrea jecorina in order to provide better insight into the possible catalytic events in these domains. The 3D structure of NAD(+)-dependent LAD1 was developed based on the crystal structure of human sorbitol dehydrogenase as a template. A series of molecular mechanics and dynamics operations were performed to find the most stable binding interaction for the enzyme and its ligands. Using the verified model, a docking study was performed with the substrate L-arabinitol, Zn(2+) and NAD(+). This study found a catalytic Zn(2+) binding domain (Cys66, His91, Glu92 and Glu176) and a cofactor NAD(+) binding domain (Gly202, ILeu204, Gly205, Cys273, Arg229 and Val298) with strong hydrogen bonding contacts with the substrate and cofactor. The binding pockets of the enzyme for l-arabinitol, NAD(+), and Zn(2+) have been explicitly defined. The results from this study should guide future mutagenesis studies and provide useful clues for engineering enzymes to improve the utilization of polyols for rare sugar production.
Collapse
Affiliation(s)
- Manish Tiwari
- Department of Chemical Engineering, Konkuk University, Seoul 143-701, Republic of Korea
| | | |
Collapse
|
43
|
Monti E, Bonten; E, D'Azzo A, Bresciani R, Venerando B, Borsani G, Schauer R, Tettamanti G. Sialidases in Vertebrates. Adv Carbohydr Chem Biochem 2010; 64:403-79. [DOI: 10.1016/s0065-2318(10)64007-3] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
44
|
Magesh S, Moriya S, Suzuki T, Miyagi T, Ishida H, Kiso M. Use of structure-based virtual screening in the investigation of novel human sialidase inhibitors. Med Chem Res 2009. [DOI: 10.1007/s00044-009-9269-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
45
|
Bigi A, Morosi L, Pozzi C, Forcella M, Tettamanti G, Venerando B, Monti E, Fusi P. Human sialidase NEU4 long and short are extrinsic proteins bound to outer mitochondrial membrane and the endoplasmic reticulum, respectively. Glycobiology 2009; 20:148-57. [PMID: 19797320 DOI: 10.1093/glycob/cwp156] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Sialidases are widely distributed glycohydrolytic enzymes removing sialic acid residues from glycoconjugates. In mammals, several sialidases with different subcellular localizations and biochemical features have been described. NEU4, the most recently identified member of the human sialidase family, is found in two forms, NEU4 long and NEU4 short, differing in the presence of a 12-amino-acid sequence at the N-terminus. Contradictory data are present in the literature about the subcellular distribution of these enzymes, their membrane anchoring mechanism being still unclear. In this work, we investigate the human NEU4 long and NEU4 short membrane anchoring mechanism and their subcellular localization. Protein extraction with Triton X-114 and sodium carbonate and cross-linking experiments demonstrate that both forms of NEU4 are extrinsic membrane proteins, anchored via protein-protein interactions. Moreover, through confocal microscopy and subcellular fractionation, we show that the long form localizes in mitochondria, while the short form is also associated with the endoplasmic reticulum. Finally, mitochondria subfractionation experiments suggest that NEU4 long is bound to the outer mitochondrial membrane.
Collapse
Affiliation(s)
- Alessandra Bigi
- Department of Biotechnologies and Biosciences, University of Milan-Bicocca, Piazza della Scienza, 2, 20126, Milano
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Magesh S, Savita V, Moriya S, Suzuki T, Miyagi T, Ishida H, Kiso M. Human sialidase inhibitors: Design, synthesis, and biological evaluation of 4-acetamido-5-acylamido-2-fluoro benzoic acids. Bioorg Med Chem 2009; 17:4595-603. [DOI: 10.1016/j.bmc.2009.04.065] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2009] [Revised: 04/28/2009] [Accepted: 04/29/2009] [Indexed: 10/20/2022]
|
47
|
Shiozaki K, Koseki K, Yamaguchi K, Shiozaki M, Narimatsu H, Miyagi T. Developmental change of sialidase neu4 expression in murine brain and its involvement in the regulation of neuronal cell differentiation. J Biol Chem 2009; 284:21157-64. [PMID: 19506080 DOI: 10.1074/jbc.m109.012708] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Sialidase Neu4 is reported to be dominantly expressed in the mouse brain, but its functional significance is not fully understood. We previously demonstrated that sialidase Neu3, also rich in mouse brain, is up-regulated during neuronal differentiation with involvement in acceleration of neurite formation. To elucidate physiological functions of Neu4, as well as Neu3, we determined expression during mouse brain development by quantitative RT-PCR. Expression was relatively low in the embryonic stage and then rapidly increased at 3-14 days after birth, whereas Neu3 demonstrated high levels in the embryonic stage and down-regulation after birth. Murine Neu4 was found to possess two isoforms differing in expression levels, developmental pattern, and enzymatic character. Distinct from the human isoforms, the murine forms, to a different extent, both catalyzed the removal of sialic acid from gangliosides as well as glycoproteins, and one isoform seemed to act on polysialylated NCAM efficiently, despite the low activity toward ordinary substrates. In situ hybridization demonstrated Neu4 mRNA to be present mainly in the hippocampus in which NCAM is rich and decreases after birth. During retinoic acid-induced differentiation, Neu4 expression was down-regulated in Neuro2a cells. Overexpression of Neu4 resulted in suppression of neurite formation, and its knockdown showed the acceleration. Thin layer chromatography of the glycolipids from Neu4-transfected cells showed ganglioside compositions to be only slightly affected, although lectin blot analysis revealed increased binding to Ricinus communis agglutinin (RCA) lectin of a approximately 95-kDa glycoprotein, which decreased with cell differentiation. These results suggest that mouse Neu4 plays an important regulatory role in neurite formation, possibly through desialylation of glycoproteins.
Collapse
Affiliation(s)
- Kazuhiro Shiozaki
- Division of Biochemistry, Miyagi Cancer Center Research Institute, Natori, Miyagi 981-1293, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Toovey S, Rayner C, Prinssen E, Chu T, Donner B, Thakrar B, Dutkowski R, Hoffmann G, Breidenbach A, Lindemann L, Carey E, Boak L, Gieschke R, Sacks S, Solsky J, Small I, Reddy D. Assessment of neuropsychiatric adverse events in influenza patients treated with oseltamivir: a comprehensive review. Drug Saf 2009; 31:1097-114. [PMID: 19026027 DOI: 10.2165/0002018-200831120-00006] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
After reports from Japan of neuropsychiatric adverse events (NPAEs) in children taking oseltamivir phosphate (hereafter referred to as oseltamivir [Tamiflu; F. Hoffmann-La Roche Ltd, Basel, Switzerland]) during and after the 2004--5 influenza season, Roche explored possible reasons for the increase in reporting rate and presented regular updates to the US FDA and other regulatory authorities. This review summarizes the results of a comprehensive assessment of the company's own preclinical and clinical studies, post-marketing spontaneous adverse event reporting, epidemiological investigations utilizing health claims and medical records databases and an extensive review of the literature, with the aim of answering the following questions: (i) what the types and rates of neuropsychiatric abnormalities reported in patients with influenza are, and whether these differ in patients who have received oseltamivir compared with those who have not; (ii) what levels of oseltamivir and its active metabolite, oseltamivir carboxylate are achieved in the CNS; (iii) whether oseltamivir and oseltamivir carboxylate have pharmacological activity in the CNS; and (iv) whether there are genetic differences between Japanese and Caucasian patients that result in different levels of oseltamivir and/or oseltamivir carboxylate in the CNS, differences in their metabolism or differences in their pharmacological activity in the CNS. In total, 3051 spontaneous reports of NPAEs were received by Roche, involving 2466 patients who received oseltamivir between 1999 and 15 September 2007; 2772 (90.9%) events originated from Japan, 190 (6.2%) from the US and 89 (2.9%) from other countries. During this period, oseltamivir was prescribed to around 48 million people worldwide. Crude NPAE reporting rates (per 1,000,000 prescriptions) in children (aged < or =16 years) and adults, respectively, were 99 and 28 events in Japan and 19 and 8 in the US. NPAEs were more commonly reported in children (2218 events in 1808 children aged < or =16 years vs 833 in 658 adults) and generally occurred within 48 hours of the onset of influenza illness and initiation of treatment. After categorizing the reported events according to International Classification of Diseases (9th edition) codes, abnormal behaviour (1160 events, 38.0%) and delusions/perceptual disturbances (661 events, 21.7%) were the largest categories of events, and delirium or delirium-like events (as defined by the American Psychiatric Association) were very common in most categories. No difference in NPAE reporting rates between oseltamivir and placebo was found in phase III treatment studies (0.5% vs 0.6%). Analyses of US healthcare claims databases showed the risk of NPAEs in oseltamivir-treated patients (n = 159,386) was no higher than those not receiving antivirals (n = 159,386). Analysis of medical records in the UK General Practice Research Database showed that the adjusted relative risk of NPAEs in influenza patients was significantly higher (1.75-fold) than in the general population. Based on literature reports, NPAEs in Japanese and Taiwanese children with influenza have occurred before the initiation of oseltamivir treatment; events were also similar to those occurring after the initiation of oseltamivir therapy. No clinically relevant differences in plasma pharmacokinetics of oseltamivir and its active metabolite oseltamivir carboxylate were noted between Japanese and Caucasian adults or children. Penetration into the CNS of both oseltamivir and oseltamivir carboxylate was low in Japanese and Caucasian adults (cerebrospinal fluid/plasma maximum concentration and area under the plasma concentration-time curve ratios of approximately 0.03), and the capacity for converting oseltamivir to oseltamivir carboxylate in rat and human brains was low. In animal autoradiography and pharmacokinetic studies, brain : plasma radioactivity ratios were generally 20% or lower. Animal studies showed no specific CNS/behavioural effects after administration of doses corresponding to > or =100 times the clinical dose. Oseltamivir or oseltamivir carboxylate did not interact with human neuraminidases or with 155 known molecular targets in radioligand binding and functional assays. A review of the information published to date on functional variations of genes relevant to oseltamivir pharmacokinetics and pharmacodynamics and simulated gene knock-out scenarios did not identify any plausible genetic explanations for the observed NPAEs. The available data do not suggest that the incidence of NPAEs in influenza patients receiving oseltamivir is higher than in those who do not, and no mechanism by which oseltamivir or oseltamivir carboxylate could cause or worsen such events could be identified.
Collapse
|
49
|
Curtis MA, Monzo HJ, Faull RL. The rostral migratory stream and olfactory system: smell, disease and slippery cells. PROGRESS IN BRAIN RESEARCH 2009; 175:33-42. [DOI: 10.1016/s0079-6123(09)17503-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
50
|
Buschiazzo A, Alzari PM. Structural insights into sialic acid enzymology. Curr Opin Chem Biol 2008; 12:565-72. [DOI: 10.1016/j.cbpa.2008.06.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Accepted: 06/17/2008] [Indexed: 01/27/2023]
|